1
|
Bravo-Vázquez LA, Mora-Hernández EO, Rodríguez AL, Sahare P, Bandyopadhyay A, Duttaroy AK, Paul S. Current Advances of Plant-Based Vaccines for Neurodegenerative Diseases. Pharmaceutics 2023; 15:711. [PMID: 36840033 PMCID: PMC9963606 DOI: 10.3390/pharmaceutics15020711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are characterized by the progressive degeneration and/or loss of neurons belonging to the central nervous system, and represent one of the major global health issues. Therefore, a number of immunotherapeutic approaches targeting the non-functional or toxic proteins that induce neurodegeneration in NDDs have been designed in the last decades. In this context, due to unprecedented advances in genetic engineering techniques and molecular farming technology, pioneering plant-based immunogenic antigen expression systems have been developed aiming to offer reliable alternatives to deal with important NDDs, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Diverse reports have evidenced that plant-made vaccines trigger significant immune responses in model animals, supported by the production of antibodies against the aberrant proteins expressed in the aforementioned NDDs. Moreover, these immunogenic tools have various advantages that make them a viable alternative for preventing and treating NDDs, such as high scalability, no risk of contamination with human pathogens, cold chain free production, and lower production costs. Hence, this article presents an overview of the current progress on plant-manufactured vaccines for NDDs and discusses its future prospects.
Collapse
Affiliation(s)
- Luis Alberto Bravo-Vázquez
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| | - Erick Octavio Mora-Hernández
- School of Engineering and Sciences, Campus Mexico City, Tecnologico de Monterrey, Calle del Puente, No. 222 Col. Ejidos de Huipulco, Tlalpan, Mexico City 14380, Mexico
| | - Alma L. Rodríguez
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| | - Padmavati Sahare
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM 3001, Juriquilla, Querétaro 76230, Mexico
| | - Anindya Bandyopadhyay
- International Rice Research Institute, Manila 4031, Philippines
- Reliance Industries Ltd., Navi Mumbai 400701, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, P.O. Box 1046 Blindern, 0317 Oslo, Norway
| | - Sujay Paul
- School of Engineering and Sciences, Campus Querétaro, Tecnologico de Monterrey, Av. Epigmenio González, No. 500 Fracc. San Pablo, Querétaro 76130, Mexico
| |
Collapse
|
2
|
Zhang L, Ma H, Wan S, Zhang Y, Gao M, Liu X. Mycobacterium tuberculosis latency-associated antigen Rv1733c SLP improves the accuracy of differential diagnosis of active tuberculosis and latent tuberculosis infection. Chin Med J (Engl) 2022; 135:63-69. [PMID: 34802023 PMCID: PMC8850866 DOI: 10.1097/cm9.0000000000001858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Differential diagnosis of active tuberculosis (ATB) and latent tuberculosis infection (LTBI) has been a challenge for clinicians in high TB burden countries. The purpose of this study was to improve the accuracy of differential diagnosis of ATB and LTBI by using fluorescent immunospot (FluoroSpot) assay to detect specific Th1 cell immune responses. The novel mycobacterium tuberculosis (MTB) latency-associated antigens Rv1733c and synthetic long peptides derived from Rv1733c (Rv1733c SLP) were used based on virulence factors early secreting antigen target-6 (ESAT-6) and culture filtrate protein-10 (CFP-10). METHODS Fifty-seven ATB cases, including 20 pathogen-confirmed ATB and 37 clinically diagnosed ATB, and 36 LTBI cases, were enrolled between January and December 2017. FluoroSpot assay was used to detect the interferon γ (IFN-γ) and interleukin 2 (IL-2) secreted by the specific T cells after being stimulated with MTB virulence factors ESAT-6 and CFP-10, MTB latency-associated antigens Rv1733c and Rv1733c SLP. The receiver operating characteristic (ROC) curve was used to define the best cutoff value of latency-associated antigens in the use of differentiating ATB and LTBI. The sensitivity, specificity, predictive value, and likelihood ratio of ESAT-6 and CFP-10-FluoroSpot combined with latency-associated antigen in the differential diagnosis of ATB and LTBI were also calculated. RESULTS Following the stimulation with Rv1733c and Rv1733c SLP, the frequency of single IL-2-secreting T cells stimulated by Rv1733c SLP had the largest area under the ROC curve, which was 0.766. With a cutoff value of 1 (spot-forming cells [SFCs]/2.5 × 105 peripheral blood mononuclear cells) for frequency, the sensitivity and specificity of distinguishing ATB from LTBI were 72.2% and 73.7%, respectively. ESAT-6 and CFP-10-FluoroSpot detected the frequency and proportion of single IFN-γ-secreting T cells; the sensitivity and specificity of distinguishing ATB from LTBI were 82.5% and 66.7%, respectively. Combined with the frequency of single IL-2-secreting T cells stimulated by Rv1733c SLP on the basis of ESAT-6 and CFP-10-FluoroSpot, the sensitivity and specificity increased to 84.2% and 83.3%, respectively. CONCLUSION Rv1733c SLP, combined with ESAT-6 and CFP-10, might be used as a candidate antigen for T cell-based tuberculosis diagnostic tests to differentiate ATB from LTBI.
Collapse
Affiliation(s)
- Lifan Zhang
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Huimin Ma
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shijun Wan
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yueqiu Zhang
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mengqiu Gao
- Department of Tuberculosis, Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing 101149, China
| | - Xiaoqing Liu
- Division of Infectious Diseases, Department of Internal Medicine, State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
- Clinical Epidemiology Unit, Peking Union Medical College, International Clinical Epidemiology Network, Beijing 100730, China
- Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
3
|
Ashi MO, Mami-Chouaib F, Corgnac S. Mutant and non-mutant neoantigen-based cancer vaccines: recent advances and future promises. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:746-762. [PMID: 36654823 PMCID: PMC9834040 DOI: 10.37349/etat.2022.00111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
Major advances in cancer treatment have emerged with the introduction of immunotherapies using blocking antibodies that target T-cell inhibitory receptors, such as programmed death-1 (PD-1) and cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), known as immune checkpoints. However, most cancer patients do not respond to immune checkpoint blockade (ICB) therapies, suggesting the development of resistance mechanisms associated with either an insufficient number of preexisting tumor-specific T-cell precursors and/or inappropriate T-cell reactivation. To broaden clinical benefit, anti-PD-1/PD-1 ligand (PD-L1) neutralizing antibodies have been combined with therapeutic cancer vaccines based on non-mutant and/or mutant tumor antigens, to stimulate and expand tumor-specific T lymphocytes. Although these combination treatments achieve the expected goal in some patients, relapse linked to alterations in antigen presentation machinery (APM) of cancer cells often occurs leading to tumor escape from CD8 T-cell immunity. Remarkably, an alternative antigenic peptide repertoire, referred to as T-cell epitopes associated with impaired peptide processing (TEIPP), arises on these malignant cells with altered APM. TEIPP are derived from ubiquitous non-mutant self-proteins and represent a unique resource to target immune-edited tumors that have acquired resistance to cytotoxic T lymphocytes (CTLs) related to defects in transporter associated with antigen processing (TAP) and possibly also to ICB. The present review discusses tumor-associated antigens (TAAs) and mutant neoantigens and their use as targets in peptide- and RNA-based therapeutic cancer vaccines. Finally, this paper highlights TEIPP as a promising immunogenic non-mutant neoantigen candidates for active cancer immunotherapy and combination with TAA and mutant neoantigens. Combining these polyepitope cancer vaccines with ICB would broaden T-cell specificity and reinvigorate exhausted antitumor CTL, resulting in the eradication of all types of neoplastic cells, including immune-escaped subtypes.
Collapse
Affiliation(s)
- Mohamad Omar Ashi
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France,Correspondence: Fathia Mami-Chouaib,
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France,Stéphanie Corgnac, . INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, 94805 Villejuif, France
| |
Collapse
|
4
|
Ahmed MM, Sayed AM, El Abd D, Fares S, Said MSM, Elsayed Sedik Ebrahim E. Gender Difference in Perceived Symptoms and Laboratory Investigations in Suspected and Confirmed COVID-19 Cases: A Retrospective Study. J Prim Care Community Health 2021; 12:21501327211039718. [PMID: 34407661 PMCID: PMC8381412 DOI: 10.1177/21501327211039718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Evaluating gender-specific effects of COVID-19 is important to develop effective therapeutic strategies. The aim of this study was to explore gender difference in perceived symptoms and laboratory investigations in suspected and confirmed cases. METHODS This is a retrospective study that included data from suspected COVID-19 patients during the first wave of the pandemic. Participants using the phone triaging system at Kasralainy outpatient clinics were included. The analyzed data included patient history and results of nasopharyngeal swab and laboratory data. RESULTS Out of 440 COVID-19 suspected cases, 56.36% were females. The perceived COVID-19 symptoms showed no significant gender difference in suspected cases while in confirmed cases females were 4 times more likely to complain of cough [OR (95% CI) 3.92 (1.316-11.68), P-value .014] and 5 times more likely to experience loss of smell or taste [OR (95% CI) 4.84 (1.62-14.43), P-value .005]. Laboratory markers revealed high levels of aspartate aminotransferase, alanine aminotransferase, blood urea, serum creatinine, creatine kinase, and serum ferritin in males and this was statistically significant (P-value <.001) in suspected and confirmed cases. Females confirmed with COVID-19 were 80%, 97%, and 97% less likely to have high levels of ALT, creatin kinase, and serum ferritin [OR (95% CI) 0.20 (0.07-0.54), 0.07 (0.01-0.38), and 0.07 (0.01-0.90), P-value .002, .002, and .041, respectively]. CONCLUSION Gender differences were found in laboratory markers in COVID-19 suspected and confirmed cases and in perceived symptoms in confirmed cases.
Collapse
Affiliation(s)
| | - Amal M Sayed
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Dina El Abd
- Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Samar Fares
- Family Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
5
|
Engelhard VH, Obeng RC, Cummings KL, Petroni GR, Ambakhutwala AL, Chianese-Bullock KA, Smith KT, Lulu A, Varhegyi N, Smolkin ME, Myers P, Mahoney KE, Shabanowitz J, Buettner N, Hall EH, Haden K, Cobbold M, Hunt DF, Weiss G, Gaughan E, Slingluff CL. MHC-restricted phosphopeptide antigens: preclinical validation and first-in-humans clinical trial in participants with high-risk melanoma. J Immunother Cancer 2021; 8:jitc-2019-000262. [PMID: 32385144 PMCID: PMC7228659 DOI: 10.1136/jitc-2019-000262] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Phosphorylated peptides presented by MHC molecules represent a new class of neoantigens expressed on cancer cells and recognized by CD8 T-cells. These peptides are promising targets for cancer immunotherapy. Previous work identified an HLA-A*0201-restricted phosphopeptide from insulin receptor substrate 2 (pIRS2) as one such target. The purpose of this study was to characterize a second phosphopeptide, from breast cancer antiestrogen resistance 3 (BCAR3), and to evaluate safety and immunogenicity of a novel immunotherapic vaccine comprising either or both of these phosphorylated peptides. METHODS Phosphorylated BCAR3 protein was evaluated in melanoma and breast cancer cell lines by Western blot, and recognition by T-cells specific for HLA-A*0201-restricted phosphorylated BCAR3 peptide (pBCAR3126-134) was determined by 51Cr release assay and intracellular cytokine staining. Human tumor explants were also evaluated by mass spectrometry for presentation of pIRS2 and pBCAR3 peptides. For the clinical trial, participants with resected stage IIA-IV melanoma were vaccinated 6 times over 12 weeks with one or both peptides in incomplete Freund's adjuvant and Hiltonol (poly-ICLC). Adverse events (AEs) were coded based on National Cancer Institute (NCI) Common Terminology Criteria for Adverse Events (CTCAE) V.4.03, with provision for early study termination if dose-limiting toxicity (DLT) rates exceeded 33%. The enrollment target was 12 participants evaluable for immune response to each peptide. T-cell responses were assessed by interferon-γ ELISpot assay. RESULTS pBCAR3 peptides were immunogenic in vivo in mice, and in vitro in normal human donors, and T-cells specific for pBCAR3126-134 controlled outgrowth of a tumor xenograft. The pIRS21097-1105 peptide was identified by mass spectrometry from human hepatocellular carcinoma tumors. In the clinical trial, 15 participants were enrolled. All had grade 1 or 2 treatment-related AEs, but there were no grade 3-4 AEs, DLTs or deaths on study. T-cell responses were induced to the pIRS21097-1105 peptide in 5/12 patients (42%, 90% CI 18% to 68%) and to the pBCAR3126-134 peptide in 2/12 patients (17%, 90% CI 3% to 44%). CONCLUSION This study supports the safety and immunogenicity of vaccines containing the cancer-associated phosphopeptides pBCAR3126-134 and pIRS21097-1105, and the data support continued development of immune therapy targeting phosphopeptides. Future studies will define ways to further enhance the magnitude and durability of phosphopeptide-specific immune responses. TRIAL REGISTRATION NUMBER NCT01846143.
Collapse
Affiliation(s)
- Victor H Engelhard
- Beirne Carter Center for Immunology Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Rebecca C Obeng
- Beirne Carter Center for Immunology Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kara L Cummings
- Beirne Carter Center for Immunology Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Gina R Petroni
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Angela L Ambakhutwala
- Beirne Carter Center for Immunology Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery/Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kelly T Smith
- Department of Surgery/Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Amanda Lulu
- Beirne Carter Center for Immunology Research and Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Nikole Varhegyi
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark E Smolkin
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Paisley Myers
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Keira E Mahoney
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Nico Buettner
- 7Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Emily H Hall
- Office of Clinical Research, University Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kathleen Haden
- Department of Surgery/Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Mark Cobbold
- 7Medical Research Council Centre for Immune Regulation and Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Geoffrey Weiss
- Medicine/Division of Hematology-Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Elizabeth Gaughan
- Medicine/Division of Hematology-Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
6
|
Lampis A, Ratti M, Ghidini M, Mirchev MB, Okuducu AF, Valeri N, Hahne JC. Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). Int J Mol Med 2021; 47:97. [PMID: 33846775 PMCID: PMC8041478 DOI: 10.3892/ijmm.2021.4930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Oesophageal cancer is one of the most aggressive malignancies with limited treatment options, thus resulting in a high morbidity and mortality. With 5‑year survival rates of only 5‑10%, oesophageal cancer holds a dismal prognosis for patients. In order to improve overall survival, the early diagnosis and tools for patient stratification for personalized treatment are urgent needs. A minority of oesophageal cancers belong to the spectrum of Lynch syndrome‑associated cancers and are characterized by microsatellite instability (MSI). Microsatellite instability is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumours, such as colorectal and gastric cancer. In the latter, high levels of MSI are associated with a better prognosis and with an increased benefit to immune‑based therapies. Therefore, similar therapeutic approaches could offer an opportunity of treatment for oesophageal cancer patients with MSI. Apart from immune checkpoint inhibitors, other immunotherapies such as adoptive T‑cell transfer, peptide vaccine and oncolytic viruses are under investigation in oesophageal cancer patients. In the present review, the rationale and current knowledge about immunotherapies in oesophageal cancer are summarised.
Collapse
Affiliation(s)
- Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| | - Margherita Ratti
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Medical Department, Division of Oncology, Hospital Trust of Cremona, I-26100 Cremona, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Hospital Policlinic 'Fondazione IRCCS Ca' Granda Ospedale Maggiore', I-20122 Milan, Italy
| | - Milko B. Mirchev
- Clinic of Gastroenterology, Medical University, 9002 Varna, Bulgaria
| | | | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, Sutton SM25NG, UK
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| |
Collapse
|
7
|
Rabaan AA, Al-Ahmed SH, Garout MA, Al-Qaaneh AM, Sule AA, Tirupathi R, Mutair AA, Alhumaid S, Hasan A, Dhawan M, Tiwari R, Sharun K, Mohapatra RK, Mitra S, Emran TB, Bilal M, Singh R, Alyami SA, Moni MA, Dhama K. Diverse Immunological Factors Influencing Pathogenesis in Patients with COVID-19: A Review on Viral Dissemination, Immunotherapeutic Options to Counter Cytokine Storm and Inflammatory Responses. Pathogens 2021; 10:565. [PMID: 34066983 PMCID: PMC8150955 DOI: 10.3390/pathogens10050565] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/02/2021] [Accepted: 05/05/2021] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is still not fully unraveled. Though preventive vaccines and treatment methods are out on the market, a specific cure for the disease has not been discovered. Recent investigations and research studies primarily focus on the immunopathology of the disease. A healthy immune system responds immediately after viral entry, causing immediate viral annihilation and recovery. However, an impaired immune system causes extensive systemic damage due to an unregulated immune response characterized by the hypersecretion of chemokines and cytokines. The elevated levels of cytokine or hypercytokinemia leads to acute respiratory distress syndrome (ARDS) along with multiple organ damage. Moreover, the immune response against SARS-CoV-2 has been linked with race, gender, and age; hence, this viral infection's outcome differs among the patients. Many therapeutic strategies focusing on immunomodulation have been tested out to assuage the cytokine storm in patients with severe COVID-19. A thorough understanding of the diverse signaling pathways triggered by the SARS-CoV-2 virus is essential before contemplating relief measures. This present review explains the interrelationships of hyperinflammatory response or cytokine storm with organ damage and the disease severity. Furthermore, we have thrown light on the diverse mechanisms and risk factors that influence pathogenesis and the molecular pathways that lead to severe SARS-CoV-2 infection and multiple organ damage. Recognition of altered pathways of a dysregulated immune system can be a loophole to identify potential target markers. Identifying biomarkers in the dysregulated pathway can aid in better clinical management for patients with severe COVID-19 disease. A special focus has also been given to potent inhibitors of proinflammatory cytokines, immunomodulatory and immunotherapeutic options to ameliorate cytokine storm and inflammatory responses in patients affected with COVID-19.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia;
| | - Shamsah H. Al-Ahmed
- Specialty Paediatric Medicine, Qatif Central Hospital, Qatif 32654, Saudi Arabia;
| | - Mohammed A. Garout
- Department of Community Medicine and Health Care for Pilgrims, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Ayman M. Al-Qaaneh
- Department of Genetic Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia;
- Clinical Pharmacy Services Division, Pharmacy Services Department, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Anupam A Sule
- Department of Informatics and Outcomes, St Joseph Mercy Oakland, Pontiac, MI 48341, USA;
| | - Raghavendra Tirupathi
- Department of Medicine Keystone Health, Penn State University School of Medicine, Hershey, PA 16801, USA;
- Department of Medicine, Wellspan Chambersburg and Waynesboro (Pa.) Hospitals, Chambersburg, PA 16801, USA
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Alahsa 36342, Saudi Arabia;
- College of Nursing, Prince Nora University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Alahsa 31982, Saudi Arabia;
| | - Abdulkarim Hasan
- Department of Pathology, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt;
- Prince Mishari Bin Saud Hospital in Baljurashi, Ministry of Health, Baljurash 22888, Saudi Arabia
| | - Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana 141004, India;
- The Trafford Group of Colleges, Manchester WA14 5PQ, UK
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Uttar Pradesh Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandha Sansthan (DUVASU), Mathura 281001, India;
| | - Khan Sharun
- Division of Surgery, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India;
| | - Ranjan K. Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar 758002, India;
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian 223003, China;
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India;
| | - Salem A. Alyami
- Department of Mathematics and Statistics, Imam Mohammad Ibn Saud Islamic University, Riyadh 11432, Saudi Arabia;
| | - Mohammad Ali Moni
- WHO Collaborating Centre on eHealth, UNSW Digital Health, School of Public Health and Community Medicine, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, India;
| |
Collapse
|
8
|
Al-kuraishy HM, Al-Gareeb AI, Faidah H, Al-Maiahy TJ, Cruz-Martins N, Batiha GES. The Looming Effects of Estrogen in Covid-19: A Rocky Rollout. Front Nutr 2021; 8:649128. [PMID: 33816542 PMCID: PMC8012689 DOI: 10.3389/fnut.2021.649128] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 02/15/2021] [Indexed: 12/17/2022] Open
Abstract
In the face of the Covid-19 pandemic, an intensive number of studies have been performed to understand in a deeper way the mechanisms behind better or worse clinical outcomes. Epidemiologically, men subjects are more prone to severe acute respiratory syndrome-coronavirus type 2 (SARS-CoV-2) infections than women, with a similar scenario being also stated to the previous coronavirus diseases, namely, SARS-CoV in 2003 and Middle East Respiratory Syndrome coronavirus diseases (MERS-CoV) in 2012. In addition, and despite that aging is regarded as an independent risk factor for the severe form of the disease, even so, women protection is evident. In this way, it has been expected that sex hormones are the main determinant factors in gender differences, with the immunomodulatory effects of estrogen in different viral infections, chiefly in Covid-19, attracting more attention as it might explain the case-fatality rate and predisposition of men for Covid-19 severity. Here, we aim to provide a mini-review and an overview on the protective effects of estrogen in Covid-19. Different search strategies were performed including Scopus, Web of Science, Medline, Pubmed, and Google Scholar database to find relative studies. Findings of the present study illustrated that women have a powerful immunomodulating effect against Covid-19 through the effect of estrogen. This study illustrates that estrogens have noteworthy anti-inflammatory and immuno-modulatory effects in Covid-19. Also, estrogen hormone reduces SARS-CoV-2 infectivity through modulation of pro-inflammatory signaling pathways. This study highlighted the potential protective effect of estrogen against Covid-19 and recommended for future clinical trial and prospective studies to elucidate and confirm this protective effect.
Collapse
Affiliation(s)
- Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyiah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Therapeutic Medicine, College of Medicine, Al-Mustansiriyiah University, Baghdad, Iraq
| | - Hani Faidah
- Microbiology, Faculty of Medicine, Umm Al Qura University, Mecca, Saudi Arabia
| | - Thabat J. Al-Maiahy
- Department of Gynecology and Obstetrics, College of Medicine, Al-Mustansiriyiah University, Baghdad, Iraq
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, Porto, Portugal
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| |
Collapse
|
9
|
Irie H, Morita K, Koizumi M, Mochizuki S. Immune Responses and Antitumor Effect through Delivering to Antigen Presenting Cells by Optimized Conjugates Consisting of CpG-DNA and Antigenic Peptide. Bioconjug Chem 2020; 31:2585-2595. [PMID: 33151667 DOI: 10.1021/acs.bioconjchem.0c00523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunotherapy using antigen-specific cytotoxic T lymphocytes (CTLs) has become one of the most attractive strategies for cancer treatment. For the induction of antigen-specific CTLs in vivo, the co-delivery of CpG-DNAs and antigens to the same antigen-presenting cells (APCs) is a promising strategy. In this study, we prepared conjugates consisting of 40mer of CpG-DNA (CpG40) and antigenic peptide (OVA257-264), which have the following distinctive features: (1) multiple CpG motifs in a molecule; (2) cleavage in the cytosol because of the disulfide bonding via cysteine residue between peptide and CpG-DNA; (3) conjugation designed to induce antigen presentation on MHC class I molecules. Immunization with the conjugate CpG40-C-OVA257-264 at the mouse tail base induced strong CTL activity at a very low peptide dose of 20 ng/head. It was found that the conjugates were internalized into C-type mannose receptor 1 (MRC1)-expressing cells in inguinal lymph nodes, indicating that the CpG portion in the conjugate acts as not only an adjuvant for the activation of TLR9 but also a carrier to APCs expressing MRC1. In a tumor-bearing mice model, mice immunized with CpG40-C-OVA257-264 conjugates exhibited long delays in tumor growth compared with those treated with PBS, OVA257-264 alone, or a mixture of CpG40 and OVA257-264. Therefore, CpG-C-peptide conjugates could be a new and effective platform for peptide vaccine for the treatment of cancers and infectious diseases.
Collapse
Affiliation(s)
- Hitomi Irie
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| | - Koji Morita
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Makoto Koizumi
- Modality Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka 808-0135, Japan
| |
Collapse
|
10
|
Agrawal H, Das N, Nathani S, Saha S, Saini S, Kakar SS, Roy P. An Assessment on Impact of COVID-19 Infection in a Gender Specific Manner. Stem Cell Rev Rep 2020; 17:94-112. [PMID: 33029768 PMCID: PMC7541100 DOI: 10.1007/s12015-020-10048-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2020] [Indexed: 12/19/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by novel coronavirus Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It was first time reported in December 2019 in Wuhan, China and thereafter quickly spread across the globe. Till September 19, 2020, COVID-19 has spread to 216 countries and territories. Severe infection of SARS-CoV-2 cause extreme increase in inflammatory chemokines and cytokines that may lead to multi-organ damage and respiratory failure. Currently, no specific treatment and authorized vaccines are available for its treatment. Renin angiotensin system holds a promising role in human physiological system specifically in regulation of blood pressure and electrolyte and fluid balance. SARS-CoV-2 interacts with Renin angiotensin system by utilizing angiotensin-converting enzyme 2 (ACE2) as a receptor for its cellular entry. This interaction hampers the protective action of ACE2 in the cells and causes injuries to organs due to persistent angiotensin II (Ang-II) level. Patients with certain comorbidities like hypertension, diabetes, and cardiovascular disease are under the high risk of COVID-19 infection and mortality. Moreover, evidence obtained from several reports also suggests higher susceptibility of male patients for COVID-19 mortality and other acute viral infections compared to females. Analysis of severe acute respiratory syndrome coronavirus (SARS) and Middle East respiratory syndrome coronavirus (MERS) epidemiological data also indicate a gender-based preference in disease consequences. The current review addresses the possible mechanisms responsible for higher COVID-19 mortality among male patients. The major underlying aspects that was looked into includes smoking, genetic factors, and the impact of reproductive hormones on immune systems and inflammatory responses. Detailed investigations of this gender disparity could provide insight into the development of patient tailored therapeutic approach which would be helpful in improving the poor outcomes of COVID-19. Graphical abstract.
Collapse
Affiliation(s)
- Himanshu Agrawal
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Neeladrisingha Das
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Sandip Nathani
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Sarama Saha
- Department of Biochemistry, All India Institute of Medical Sciences, Rishikesh, India
| | - Surendra Saini
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Sham S Kakar
- Department of Physiology, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40292, USA
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
11
|
Engineering anti-cancer nanovaccine based on antigen cross-presentation. Biosci Rep 2020; 39:220729. [PMID: 31652460 PMCID: PMC6822533 DOI: 10.1042/bsr20193220] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DCs) present exogenous antigens on major histocompatibility complex (MHC) class I molecules, thereby activating CD8+ T cells, contributing to tumor elimination through a mechanism known as antigen cross-presentation. A variety of factors such as maturation state of DCs, co-stimulatory signals, T-cell microenvironment, antigen internalization routes and adjuvants regulate the process of DC-mediated antigen cross-presentation. Recently, the development of successful cancer immunotherapies may be attributed to the ability of DCs to cross-present tumor antigens. In this review article, we focus on the underlying mechanism of antigen cross-presentation and ways to improve antigen cross-presentation in different DC subsets. We have critically summarized the recent developments in the generation of novel nanovaccines for robust CD8+ T-cell response in cancer. In this context, we have reviewed nanocarriers that have been used for cancer immunotherapeutics based on antigen cross-presentation mechanism. Additionally, we have also expressed our views on the future applications of this mechanism in curing cancer.
Collapse
|
12
|
Schizas D, Charalampakis N, Kole C, Mylonas KS, Katsaros I, Zhao M, Ajani JA, Psyrri A, Karamouzis MV, Liakakos T. Immunotherapy for esophageal cancer: a 2019 update. Immunotherapy 2020; 12:203-218. [PMID: 32208794 DOI: 10.2217/imt-2019-0153] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Esophageal cancer remains a global health concern with a dismal prognosis and an estimated 5-year survival rate of approximately 10-15%. Immunotherapy is a novel treatment approach representing an effective and promising option against several types of cancer. The development of new and efficacious immunotherapeutic strategies, such as adoptive cell therapy-based, antibody-based and vaccine-based therapies, aims to prevent immunological escape and modify immunological responses. In this review, we discuss the theoretical background and current status of immunotherapy for patients with esophageal cancer. We also present ongoing clinical trials and summarize key findings concerning survival and safety analyses.
Collapse
Affiliation(s)
- Dimitrios Schizas
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | | | - Christo Kole
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Konstantinos S Mylonas
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Ioannis Katsaros
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Meina Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Amanda Psyrri
- Department of Internal Medicine, Section of Medical Oncology, National & Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, National & Kapodistrian University of Athens, Athens, Greece
| | - Theodore Liakakos
- First Department of Surgery, National & Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| |
Collapse
|
13
|
Onodi F, Maherzi-Mechalikh C, Mougel A, Ben Hamouda N, Taboas C, Gueugnon F, Tran T, Nozach H, Marcon E, Gey A, Terme M, Bouzidi A, Maillere B, Kerzerho J, Tartour E, Tanchot C. High Therapeutic Efficacy of a New Survivin LSP-Cancer Vaccine Containing CD4 + and CD8 + T-Cell Epitopes. Front Oncol 2018; 8:517. [PMID: 30483475 PMCID: PMC6243131 DOI: 10.3389/fonc.2018.00517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
The efficacy of an antitumoral vaccine relies both on the choice of the antigen targeted and on its design. The tumor antigen survivin is an attractive target to develop therapeutic cancer vaccines because of its restricted over-expression and vital functions in most human tumors. Accordingly, several clinical trials targeting survivin in various cancer indications have been conducted. Most of them relied on short peptide-based vaccines and showed promising, but limited clinical results. In this study, we investigated the immunogenicity and therapeutic efficacy of a new long synthetic peptide (LSP)-based cancer vaccine targeting the tumor antigen survivin (SVX). This SVX vaccine is composed of three long synthetic peptides containing several CD4+ and CD8+ T-cell epitopes, which bind to various HLA class II and class I molecules. Studies in healthy individuals showed CD4+ and CD8+ T-cell immunogenicity of SVX peptides in human, irrespective of the individual's HLA types. Importantly, high frequencies of spontaneous T-cell precursors specific to SVX peptides were also detected in the blood of various cancer patients, demonstrating the absence of tolerance against these peptides. We then demonstrated SVX vaccine's high therapeutic efficacy against four different established murine tumor models, associated with its capacity to generate both specific cytotoxic CD8+ and multifunctional Th1 CD4+ T-cell responses. When tumors were eradicated, generated memory T-cell responses protected against rechallenge allowing long-term protection against relapses. Treatment with SVX vaccine was also found to reshape the tumor microenvironment by increasing the tumor infiltration of both CD4+ and CD8+ T cells but not Treg cells therefore tipping the balance toward a highly efficient immune response. These results highlight that this LSP-based SVX vaccine appears as a promising cancer vaccine and warrants its further clinical development.
Collapse
Affiliation(s)
- Fanny Onodi
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Chahrazed Maherzi-Mechalikh
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alice Mougel
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nadine Ben Hamouda
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Charlotte Taboas
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Fabien Gueugnon
- VAXEAL Research, Evry, France.,CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Thi Tran
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Herve Nozach
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Elodie Marcon
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Alain Gey
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Magali Terme
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Bernard Maillere
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | | | - Eric Tartour
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Corinne Tanchot
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| |
Collapse
|
14
|
Panahi HA, Bolhassani A, Javadi G, Noormohammadi Z. A comprehensive in silico analysis for identification of therapeutic epitopes in HPV16, 18, 31 and 45 oncoproteins. PLoS One 2018; 13:e0205933. [PMID: 30356257 PMCID: PMC6200245 DOI: 10.1371/journal.pone.0205933] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/11/2018] [Indexed: 11/25/2022] Open
Abstract
Human papillomaviruses (HPVs) are a group of circular double-stranded DNA viruses, showing severe tropism to mucosal tissues. A subset of HPVs, especially HPV16 and 18, are the primary etiological cause for several epithelial cell malignancies, causing about 5.2% of all cancers worldwide. Due to the high prevalence and mortality, HPV-associated cancers have remained as a significant health problem in human society, making an urgent need to develop an effective therapeutic vaccine against them. Achieving this goal is primarily dependent on the identification of efficient tumor-associated epitopes, inducing a robust cell-mediated immune response. Previous information has shown that E5, E6, and E7 early proteins are responsible for the induction and maintenance of HPV-associated cancers. Therefore, the prediction of major histocompatibility complex (MHC) class I T cell epitopes of HPV16, 18, 31 and 45 oncoproteins was targeted in this study. For this purpose, a two-step plan was designed to identify the most probable CD8+ T cell epitopes. In the first step, MHC-I and II binding, MHC-I processing, MHC-I population coverage and MHC-I immunogenicity prediction analyses, and in the second step, MHC-I and II protein-peptide docking, epitope conservation, and cross-reactivity with host antigens’ analyses were carried out successively by different tools. Finally, we introduced five probable CD8+ T cell epitopes for each oncoprotein of the HPV genotypes (60 epitopes in total), which obtained better scores by an integrated approach. These predicted epitopes are valuable candidates for in vitro or in vivo therapeutic vaccine studies against the HPV-associated cancers. Additionally, this two-step plan that each step includes several analyses to find appropriate epitopes provides a rational basis for DNA- or peptide-based vaccine development.
Collapse
Affiliation(s)
- Heidar Ali Panahi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
- * E-mail: ,
| | - Gholamreza Javadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zahra Noormohammadi
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
15
|
Durgeau A, Virk Y, Corgnac S, Mami-Chouaib F. Recent Advances in Targeting CD8 T-Cell Immunity for More Effective Cancer Immunotherapy. Front Immunol 2018; 9:14. [PMID: 29403496 PMCID: PMC5786548 DOI: 10.3389/fimmu.2018.00014] [Citation(s) in RCA: 322] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
Recent advances in cancer treatment have emerged from new immunotherapies targeting T-cell inhibitory receptors, including cytotoxic T-lymphocyte associated antigen (CTLA)-4 and programmed cell death (PD)-1. In this context, anti-CTLA-4 and anti-PD-1 monoclonal antibodies have demonstrated survival benefits in numerous cancers, including melanoma and non-small-cell lung carcinoma. PD-1-expressing CD8+ T lymphocytes appear to play a major role in the response to these immune checkpoint inhibitors (ICI). Cytotoxic T lymphocytes (CTL) eliminate malignant cells through recognition by the T-cell receptor (TCR) of specific antigenic peptides presented on the surface of cancer cells by major histocompatibility complex class I/beta-2-microglobulin complexes, and through killing of target cells, mainly by releasing the content of secretory lysosomes containing perforin and granzyme B. T-cell adhesion molecules and, in particular, lymphocyte-function-associated antigen-1 and CD103 integrins, and their cognate ligands, respectively, intercellular adhesion molecule 1 and E-cadherin, on target cells, are involved in strengthening the interaction between CTL and tumor cells. Tumor-specific CTL have been isolated from tumor-infiltrating lymphocytes and peripheral blood lymphocytes (PBL) of patients with varied cancers. TCRβ-chain gene usage indicated that CTL identified in vitro selectively expanded in vivo at the tumor site compared to autologous PBL. Moreover, functional studies indicated that these CTL mediate human leukocyte antigen class I-restricted cytotoxic activity toward autologous tumor cells. Several of them recognize truly tumor-specific antigens encoded by mutated genes, also known as neoantigens, which likely play a key role in antitumor CD8 T-cell immunity. Accordingly, it has been shown that the presence of T lymphocytes directed toward tumor neoantigens is associated with patient response to immunotherapies, including ICI, adoptive cell transfer, and dendritic cell-based vaccines. These tumor-specific mutation-derived antigens open up new perspectives for development of effective second-generation therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Aurélie Durgeau
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,ElyssaMed, Paris Biotech Santé, Paris, France
| | - Yasemin Virk
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
16
|
|
17
|
Wu Q, Pi L, Le Trinh T, Zuo C, Xia M, Jiao Y, Hou Z, Jo S, Puszyk W, Pham K, Nelson DR, Robertson K, Ostrov D, Rameshwar P, Xia CQ, Liu C. A Novel Vaccine Targeting Glypican-3 as a Treatment for Hepatocellular Carcinoma. Mol Ther 2017; 25:2299-2308. [PMID: 28865999 DOI: 10.1016/j.ymthe.2017.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has a high morbidity and mortality rate worldwide, with limited treatment options. Glypican-3 (GPC3) is a glycosylphosphatidylinositol-anchored glycoprotein that is overexpressed in most HCC tissues but not in normal tissues. GPC3-targeting antibody therapy shows limited response in a clinical trial due to the lack of a tumor-specific cytotoxic T lymphocyte (CTL) response. Here, in C57/B6 mice, we demonstrated that intravenous infusion of GPC3-coupled lymphocytes (LC/GPC3+) elicited robust GPC3-specific antibody and CTL responses, which effectively restricted proliferation and lysed cultured-HCC cells. Treatment with LC/GPC3+ induced durable tumor regression in HCC-bearing C57/B6 mice. Administration of LC/GPC3+ induced elevated levels of the cytotoxic T cell bioactive factors tumor necrosis factor alpha (TNF-α), interferon-γ (IFN-γ), granzyme B, and perforin, and substantially increased the number of infiltrating CD8+ T cells in tumor tissues. Moreover, immune responses elicited by LC/GPC3+ selectively suppressed GPC3+ tumors, but didn't affect the GPC3- tumors in BALB/c mice. Our findings provide the first preclinical evidence that intravenous infusion of the LC/GPC3+ complex can induce a strong anti-HCC effect through regulating systemic and local immune responses. These results indicate that the LC/GPC3+ complex could be developed as precision therapeutics for HCC patients in the future.
Collapse
Affiliation(s)
- Qunfeng Wu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Liya Pi
- Department of Pediatrics, University of Florida, Gainesville, FL 32611, USA
| | - Thu Le Trinh
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA; Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Chaohui Zuo
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Gastroduodenal and Pancreatic Surgery, Translational Medicine Research Center of Liver Cancer, Hunan Province Cancer Hospital and Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan Province 410013, P.R. China
| | - Man Xia
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Laboratory of Digestive Oncology, Hunan Province Cancer Institute, Changsha, Hunan Province 410013, P.R. China
| | - Yu Jiao
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32611, USA
| | - Zhouhua Hou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Sung Jo
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - William Puszyk
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Kien Pham
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA
| | - David R Nelson
- Department of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Keith Robertson
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Rochester, MN 85259, USA
| | - David Ostrov
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine, Hematology/Oncology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Chang Qing Xia
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| | - Chen Liu
- Department of Pathology and Laboratory Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA; Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
18
|
Voena C, Menotti M, Mastini C, Di Giacomo F, Longo DL, Castella B, Merlo MEB, Ambrogio C, Wang Q, Minero VG, Poggio T, Martinengo C, D'Amico L, Panizza E, Mologni L, Cavallo F, Altruda F, Butaney M, Capelletti M, Inghirami G, Jänne PA, Chiarle R. Efficacy of a Cancer Vaccine against ALK-Rearranged Lung Tumors. Cancer Immunol Res 2015; 3:1333-1343. [PMID: 26419961 DOI: 10.1158/2326-6066.cir-15-0089] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/23/2015] [Indexed: 01/14/2023]
Abstract
Non-small cell lung cancer (NSCLC) harboring chromosomal rearrangements of the anaplastic lymphoma kinase (ALK) gene is treated with ALK tyrosine kinase inhibitors (TKI), but the treatment is successful for only a limited amount of time; most patients experience a relapse due to the development of drug resistance. Here, we show that a vaccine against ALK induced a strong and specific immune response that both prophylactically and therapeutically impaired the growth of ALK-positive lung tumors in mouse models. The ALK vaccine was efficacious also in combination with ALK TKI treatment and significantly delayed tumor relapses after TKI suspension. We found that lung tumors containing ALK rearrangements induced an immunosuppressive microenvironment, regulating the expression of PD-L1 on the surface of lung tumor cells. High PD-L1 expression reduced ALK vaccine efficacy, which could be restored by administration of anti-PD-1 immunotherapy. Thus, combinations of ALK vaccine with TKIs and immune checkpoint blockade therapies might represent a powerful strategy for the treatment of ALK-driven NSCLC.
Collapse
Affiliation(s)
- Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Matteo Menotti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Cristina Mastini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Filomena Di Giacomo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Dario Livio Longo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Imaging Center, University of Torino, Torino, Italy
| | - Barbara Castella
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Maria Elena Boggio Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Chiara Ambrogio
- Molecular Oncology Program, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Qi Wang
- Department of Pathology, Children's Hospital Harvard Medical School, Boston, MA 02115, USA
| | - Valerio Giacomo Minero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Teresa Poggio
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Cinzia Martinengo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Lucia D'Amico
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Elena Panizza
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Luca Mologni
- Department of Health Sciences, University of Milano-Bicocca, Milano, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Mohit Butaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Marzia Capelletti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Giorgio Inghirami
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.,Belfer Institute for Applied Cancer Science, Dana Farber Cancer Institute, Boston, MA 02115, USA
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy.,Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza, Torino, Italy.,Department of Pathology, Children's Hospital Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
19
|
Abstract
The clinical benefit of therapeutic cancer vaccines has been established. Whereas regression of lesions was shown for premalignant lesions caused by HPV, clinical benefit in cancer patients was mostly noted as prolonged survival. Suboptimal vaccine design and an immunosuppressive cancer microenvironment are the root causes of the lack of cancer eradication. Effective cancer vaccines deliver concentrated antigen to both HLA class I and II molecules of DCs, promoting both CD4 and CD8 T cell responses. Optimal vaccine platforms include DNA and RNA vaccines and synthetic long peptides. Antigens of choice include mutant sequences, selected cancer testis antigens, and viral antigens. Drugs or physical treatments can mitigate the immunosuppressive cancer microenvironment and include chemotherapeutics, radiation, indoleamine 2,3-dioxygenase (IDO) inhibitors, inhibitors of T cell checkpoints, agonists of selected TNF receptor family members, and inhibitors of undesirable cytokines. The specificity of therapeutic vaccination combined with such immunomodulation offers an attractive avenue for the development of future cancer therapies.
Collapse
|
20
|
Synthetic Long Peptide Derived from Mycobacterium tuberculosis Latency Antigen Rv1733c Protects against Tuberculosis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015. [PMID: 26202436 DOI: 10.1128/cvi.00271-15] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Responsible for 9 million new cases of active disease and nearly 2 million deaths each year, tuberculosis (TB) remains a global health threat of overwhelming dimensions. Mycobacterium bovis BCG, the only licensed vaccine available, fails to confer lifelong protection and to prevent reactivation of latent infection. Although 15 new vaccine candidates are now in clinical trials, an effective vaccine against TB remains elusive, and new strategies for vaccination are vital. BCG vaccination fails to induce immunity against Mycobacterium tuberculosis latency antigens. Synthetic long peptides (SLPs) combined with adjuvants have been studied mostly for therapeutic cancer vaccines, yet not for TB, and proved to induce efficient antitumor immunity. This study investigated an SLP derived from Rv1733c, a major M. tuberculosis latency antigen which is highly expressed by "dormant" M. tuberculosis and well recognized by T cells from latently M. tuberculosis-infected individuals. In order to assess its in vivo immunogenicity and protective capacity, Rv1733c SLP in CpG was administered to HLA-DR3 transgenic mice. Immunization with Rv1733c SLP elicited gamma interferon-positive/tumor necrosis factor-positive (IFN-γ(+)/TNF(+)) and IFN-γ(+) CD4(+) T cells and Rv1733c-specific antibodies and led to a significant reduction in the bacterial load in the lungs of M. tuberculosis-challenged mice. This was observed both in a pre- and in a post-M. tuberculosis challenge setting. Moreover, Rv1733c SLP immunization significantly boosted the protective efficacy of BCG, demonstrating the potential of M. tuberculosis latency antigens to improve BCG efficacy. These data suggest a promising role for M. tuberculosis latency antigen Rv1733c-derived SLPs as a novel TB vaccine approach, both in a prophylactic and in a postinfection setting.
Collapse
|
21
|
Rosenthal KS, Mikecz K, Steiner HL, Glant TT, Finnegan A, Carambula RE, Zimmerman DH. Rheumatoid arthritis vaccine therapies: perspectives and lessons from therapeutic ligand epitope antigen presentation system vaccines for models of rheumatoid arthritis. Expert Rev Vaccines 2015; 14:891-908. [PMID: 25787143 DOI: 10.1586/14760584.2015.1026330] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The current status of therapeutic vaccines for autoimmune diseases is reviewed with rheumatoid arthritis as the focus. Therapeutic vaccines for autoimmune diseases must regulate or subdue responses to common self-antigens. Ideally, such a vaccine would initiate an antigen-specific modulation of the T-cell immune response that drives the inflammatory disease. Appropriate animal models and types of T helper cells and signature cytokine responses that drive autoimmune disease are also discussed. Interpretation of these animal models must be done cautiously because the means of initiation, autoantigens, and even the signature cytokine and T helper cell (Th1 or Th17) responses that are involved in the disease may differ significantly from those in humans. We describe ligand epitope antigen presentation system vaccine modulation of T-cell autoimmune responses as a strategy for the design of therapeutic vaccines for rheumatoid arthritis, which may also be effective in other autoimmune conditions.
Collapse
|
22
|
Abstract
Newer immunotherapy agents may break the barrier that tumors create to evade the attack from the immune system. Dendritic cell vaccination has shown encouraging clinical activity and a favorable safety profile in advanced tumor stages. However, optimal cell maturation status, choice of tumor antigens and route of administration have not been established. Single or multiple peptides derived from tumor-associated antigens may also be used for cancer vaccination. Intratumoral delivery of oncolytic viruses expressing immunostimulating cytokines like GM-CSF have produced stimulating clinical results that need further verification. But it is probably T-cell checkpoint modulation with monoclonal antibodies that has attracted the highest expectations. Promising activity has been reported for tremelimumab, a CTLA-4 inhibitor, and a clinical trial testing the PD-1 antibody nivolumab is underway. Future progress will probably come from a better understanding of the mechanisms of cancer-related immunosuppression, improvement in agents and strategies and combination of the available therapeutic tools.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain.,Liver Unit, Clínica Universidad de Navarra, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Avda. Pio XII 36. 31008-Pamplona, Spain
| | - Daniel Palmer
- The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK.,The Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, L69 3GA, UK
| | - Ignacio Melero
- Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain.,Departments of Oncology & Immunology, Centro de Investigación Médica Aplicada y Clínica Universidad de Navarra. Avda. Pio XII, 55. 31008-Pamplona, Spain
| |
Collapse
|
23
|
Peptide Vaccine: Progress and Challenges. Vaccines (Basel) 2014; 2:515-36. [PMID: 26344743 PMCID: PMC4494216 DOI: 10.3390/vaccines2030515] [Citation(s) in RCA: 456] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/10/2014] [Accepted: 06/13/2014] [Indexed: 12/17/2022] Open
Abstract
Conventional vaccine strategies have been highly efficacious for several decades in reducing mortality and morbidity due to infectious diseases. The bane of conventional vaccines, such as those that include whole organisms or large proteins, appear to be the inclusion of unnecessary antigenic load that, not only contributes little to the protective immune response, but complicates the situation by inducing allergenic and/or reactogenic responses. Peptide vaccines are an attractive alternative strategy that relies on usage of short peptide fragments to engineer the induction of highly targeted immune responses, consequently avoiding allergenic and/or reactogenic sequences. Conversely, peptide vaccines used in isolation are often weakly immunogenic and require particulate carriers for delivery and adjuvanting. In this article, we discuss the specific advantages and considerations in targeted induction of immune responses by peptide vaccines and progresses in the development of such vaccines against various diseases. Additionally, we also discuss the development of particulate carrier strategies and the inherent challenges with regard to safety when combining such technologies with peptide vaccines.
Collapse
|
24
|
Commandeur S, van den Eeden SJF, Dijkman K, Clark SO, van Meijgaarden KE, Wilson L, Franken KLMC, Williams A, Christensen D, Ottenhoff THM, Geluk A. The in vivo expressed Mycobacterium tuberculosis (IVE-TB) antigen Rv2034 induces CD4⁺ T-cells that protect against pulmonary infection in HLA-DR transgenic mice and guinea pigs. Vaccine 2014; 32:3580-8. [PMID: 24837764 DOI: 10.1016/j.vaccine.2014.05.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/24/2014] [Accepted: 05/01/2014] [Indexed: 12/17/2022]
Abstract
Tuberculosis (TB) remains a life-threatening infectious disease of global proportions with serious negative health and economic consequences. The lack of sufficient protection induced by Mycobacterium bovis BCG, the current vaccine for TB, as well as the impact of HIV co-infection and the emergence of drug resistant Mycobacterium tuberculosis (Mtb) strains all urge for improved vaccines against TB. A minimal requirement for Mtb vaccine antigens is their in vivo expression during Mtb infection and ability to trigger significant immune responses. Recently we identified a new class of Mtb antigens, designated IVE-TB (in vivo expressed) antigens. These included Rv2034, a protein that was expressed during pulmonary infection and strongly recognized by human T-cells. Here, the in vivo immunogenicity and protective efficacy of Rv2034 was further analyzed using HLA-DR transgenic mice that lack endogenous murine MHC class II molecules. The Rv2034 protein indeed was highly immunogenic in HLA-DR3 transgenic mice and induced HLA-DR3 restricted IFN-γ(+)/TNF(+) and IFN-γ(+) CD4(+) T-cells, specific for an epitope encoded in peptide 31-50. CD4(+) T-cell responses were optimally induced when using TLR9- and TLR3-ligand-adjuvants or CAF09. Rv2034-specific antibodies were observed following immunization with either TLR2-, TLR3-, TLR4-, TLR5-, TLR7- or TLR9-ligands or CAF09. Importantly, immunization with Rv2034 or the hybrid-protein Ag85B-ESAT6-Rv2034 adjuvanted with CpG or CAF09, induced over one log reduction, relative to unvaccinated controls, in the number of bacilli in the lungs of Mtb challenged HLA-DR3 transgenic mice and guinea pigs. These data demonstrate the potential of Rv2034 as a novel, IVE-TB antigen for future TB vaccination.
Collapse
Affiliation(s)
- Susanna Commandeur
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands
| | | | - Karin Dijkman
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands
| | - Simon O Clark
- Public Health England, Microbiology Services, Porton Down, Salisbury, Wiltshire, United Kingdom
| | | | - Louis Wilson
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands
| | - Kees L M C Franken
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands
| | - Ann Williams
- Public Health England, Microbiology Services, Porton Down, Salisbury, Wiltshire, United Kingdom
| | | | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands
| | - Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands.
| |
Collapse
|
25
|
Ménager J, Ebstein F, Oger R, Hulin P, Nedellec S, Duverger E, Lehmann A, Kloetzel PM, Jotereau F, Guilloux Y. Cross-presentation of synthetic long peptides by human dendritic cells: a process dependent on ERAD component p97/VCP but Not sec61 and/or Derlin-1. PLoS One 2014; 9:e89897. [PMID: 24587108 PMCID: PMC3937416 DOI: 10.1371/journal.pone.0089897] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/25/2014] [Indexed: 12/22/2022] Open
Abstract
Antitumor vaccination using synthetic long peptides (SLP) is an additional therapeutic strategy currently under development. It aims to activate tumor-specific CD8+ CTL by professional APCs such as DCs. DCs can activate T lymphocytes by MHC class I presentation of exogenous antigens - a process referred to as “cross-presentation”. Until recently, the intracellular mechanisms involved in cross-presentation of soluble antigens have been unclear. Here, we characterize the cross-presentation pathway of SLP Melan-A16–40 containing the HLA-A2-restricted epitope26–35 (A27L) in human DCs. Using confocal microscopy and specific inhibitors, we show that SLP16–40 is rapidly taken up by DC and follows a classical TAP- and proteasome-dependent cross-presentation pathway. Our data support a role for the ER-associated degradation machinery (ERAD)-related protein p97/VCP in the transport of SLP16–40 from early endosomes to the cytoplasm but formally exclude both sec61 and Derlin-1 as possible retro-translocation channels for cross-presentation. In addition, we show that generation of the Melan-A26–35 peptide from the SLP16–40 was absolutely not influenced by the proteasome subunit composition in DC. Altogether, our findings propose a model for cross-presentation of SLP which tends to enlarge the repertoire of potential candidates for retro-translocation of exogenous antigens to the cytosol.
Collapse
Affiliation(s)
- Jérémie Ménager
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Frédéric Ebstein
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Romain Oger
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Philippe Hulin
- INSERM U892, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | | | - Eric Duverger
- Glycobiochimie, ICOA, Université d’Orléans, Orléans, France
| | - Andrea Lehmann
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Peter-Michael Kloetzel
- Institut of Biochemistry, Charité University Hospital, Humboldt University, Berlin, Germany
| | - Francine Jotereau
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
| | - Yannick Guilloux
- INSERM U892, Nantes, France
- Université de Nantes, Nantes, France
- CNRS, UMR 6299, Nantes, France
- * E-mail:
| |
Collapse
|
26
|
Dudek AM, Martin S, Garg AD, Agostinis P. Immature, Semi-Mature, and Fully Mature Dendritic Cells: Toward a DC-Cancer Cells Interface That Augments Anticancer Immunity. Front Immunol 2013; 4:438. [PMID: 24376443 PMCID: PMC3858649 DOI: 10.3389/fimmu.2013.00438] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/23/2013] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) are the sentinel antigen-presenting cells of the immune system; such that their productive interface with the dying cancer cells is crucial for proper communication of the "non-self" status of cancer cells to the adaptive immune system. Efficiency and the ultimate success of such a communication hinges upon the maturation status of the DCs, attained following their interaction with cancer cells. Immature DCs facilitate tolerance toward cancer cells (observed for many apoptotic inducers) while fully mature DCs can strongly promote anticancer immunity if they secrete the correct combinations of cytokines [observed when DCs interact with cancer cells undergoing immunogenic cell death (ICD)]. However, an intermediate population of DC maturation, called semi-mature DCs exists, which can potentiate either tolerogenicity or pro-tumorigenic responses (as happens in the case of certain chemotherapeutics and agents exerting ambivalent immune reactions). Specific combinations of DC phenotypic markers, DC-derived cytokines/chemokines, dying cancer cell-derived danger signals, and other less characterized entities (e.g., exosomes) can define the nature and evolution of the DC maturation state. In the present review, we discuss these different maturation states of DCs, how they might be attained and which anticancer agents or cell death modalities (e.g., tolerogenic cell death vs. ICD) may regulate these states.
Collapse
Affiliation(s)
- Aleksandra M Dudek
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Shaun Martin
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Abhishek D Garg
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| | - Patrizia Agostinis
- Laboratory of Cell Death Research and Therapy, Department of Cellular and Molecular Medicine, KU Leuven , Leuven , Belgium
| |
Collapse
|
27
|
Abstract
The past decade has seen tremendous developments in novel cancer therapies through the targeting of tumor-cell-intrinsic pathways whose activity is linked to genetic alterations and the targeting of tumor-cell-extrinsic factors, such as growth factors. Furthermore, immunotherapies are entering the clinic at an unprecedented speed after the demonstration that T cells can efficiently reject tumors and that their antitumor activity can be enhanced with antibodies against immune-regulatory molecules (checkpoint blockade). Current immunotherapy strategies include monoclonal antibodies against tumor cells or immune-regulatory molecules, cell-based therapies such as adoptive transfer of ex-vivo-activated T cells and natural killer cells, and cancer vaccines. Herein, we discuss the immunological basis for therapeutic cancer vaccines and how the current understanding of dendritic cell and T cell biology might enable the development of next-generation curative therapies for individuals with cancer.
Collapse
|
28
|
Turning Tumors into Vaccines: Co-opting the Innate Immune System. Immunity 2013; 39:27-37. [DOI: 10.1016/j.immuni.2013.07.011] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/10/2013] [Indexed: 02/07/2023]
|
29
|
Geluk A, van den Eeden SJF, van Meijgaarden KE, Dijkman K, Franken KLMC, Ottenhoff THM. A multistage-polyepitope vaccine protects against Mycobacterium tuberculosis infection in HLA-DR3 transgenic mice. Vaccine 2012; 30:7513-21. [PMID: 23103299 DOI: 10.1016/j.vaccine.2012.10.045] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/03/2012] [Accepted: 10/13/2012] [Indexed: 10/27/2022]
Abstract
Mycobacterium tuberculosis (Mtb) is responsible for almost 2 million deaths annually. BCG, currently the only TB vaccine, induces variable protection and does not protect against reactivation of latent TB. Thus, efficient vaccines to supplement BCG are required urgently. Since Mtb's proteome differs qualitatively and quantitatively during bacterial replication stages from that expressed during dormancy, improved TB vaccines should drive immune responses to Mtb antigens expressed during multiple stages of infection. Consequently, such "multistage" vaccines should be composed of (immunodominant) antigens expressed during different phases of Mtb infection. As a concept multistage vaccine, we constructed a polyepitope by fusing five HLA-DR3-restricted T-cell epitopes derived from different Mtb proteins either expressed highly by replicating bacteria (Ag85B, hsp65, 19 kDa lipoprotein), or abundantly expressed by dormant bacilli and recognized preferentially by TST(+) individuals (hsp16, Rv1733c). PBMC of HLA-DR3(+) but not HLA-DR3(-) cured TB patients and TST(+) individuals responded well to the multistage-polyepitope in vitro. The in vivo immunogenicity and protective efficacy of the multistage-polyepitope were analyzed using HLA-DR3 transgenic mice lacking endogenous murine class II as a model. Immunization with the multistage-polyepitope adjuvanted with CpG generated high IgG levels as well as polyfunctional CD4(+) T-cells producing IFN-γ, TNF and IL-2, specific for these HLA-DR3-restricted epitopes. Importantly, multistage-polyepitope immunization reduced the number of bacilli in the lungs after Mtb challenge when administered as prophylactic vaccine. Given the extensive repertoire of potential Mtb antigens available for immune recognition, the data of our model demonstrate the potential of multistage-polyepitope vaccines to protect against TB.
Collapse
Affiliation(s)
- Annemieke Geluk
- Department of Infectious Diseases, Leiden University Medical Centre, The Netherlands.
| | | | | | | | | | | |
Collapse
|